Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Medicine (Baltimore) ; 102(42): e35628, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37861529

RESUMEN

This study aimed to investigate the underlying mechanism of Zuogui Pill in its efficacy against liver cancer, employing a combination of data mining approaches and network pharmacology methods. A novel clustering analysis algorithm was proposed to identify the core gene modules of Zuogui Pill. This algorithm successfully identified 5 core modules, with the first large module comprised of twelve proteins forming a 12-clique, representing the strongest connections among them. By utilizing GEO platform, ten key target proteins were detected, including FOS, PTGS2, and MYC. According to the GO annotation and KEGG analysis, desired target proteins were significantly enriched in various biological processes (BP). The analysis showed that ten key targets were strongly associated with signaling pathways mainly centered on MAPK and PI3K-Akt pathway. Additionally, molecular docking revealed strong binding affinities between core active ingredients of Zuogui Pill and these key targets, and the best affinity modes were observed for PTGS2-Sesamin, PRKCA-Sesamin, FOS-delta-Carotene. In order to establish the relationships between clinical symptoms and drug targets, a heterogeneous targets-related network was constructed. A total of 60 key target-symptom association pairs were detected, exemplified by the strongly association between fever and PTGS2 through the intermediary of Shu Di Huang. In summary, symptom-target associations are valuable in uncovering the underlying molecular mechanisms of Zuogui Pill. Our work reinforced the notion that Zuogui pill exhibits therapeutic potential on liver cancer through network targets, as well as synergistic effects of multi-component and multi-pathway. This study provided specific references for future experiments at the cost of less time.


Asunto(s)
Medicamentos Herbarios Chinos , Neoplasias Hepáticas , Humanos , Ciclooxigenasa 2 , Simulación del Acoplamiento Molecular , Farmacología en Red , Fosfatidilinositol 3-Quinasas , Neoplasias Hepáticas/tratamiento farmacológico , Aprendizaje Automático , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico
2.
Mol Neurobiol ; 60(10): 5592-5606, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37329381

RESUMEN

The transformation of microglia to a pro-inflammatory phenotype at the site of traumatic brain injury (TBI) drives the progression of secondary neurodegeneration and irreversible neurological impairment. Omega-3 polyunsaturated fatty acids (PUFA) have been shown to suppress this phenotype transformation, thereby reducing neuroinflammation following TBI, but the molecular mechanisms are unknown. We found that Omega-3 PUFA suppressed the expression of disintegrin metalloproteinase (ADAM17), the enzyme required to convert tumor necrosis factor-α (TNF-α) to the soluble form, thereby inhibiting the TNF-α/NF-κB pathway both in vitro and in a mouse model of TBI. Omega-3 PUFA also prevented the reactive transformation of microglia and promoted the secretion of microglial exosomes containing nerve growth factor (NGF), activating the neuroprotective NGF/TrkA pathway both in culture and TBI model mice. Moreover, Omega-3 PUFA suppressed the pro-apoptotic NGF/P75NTR pathway at the TBI site and reduced apoptotic neuronal death, brain edema, and disruption of the blood-brain barrier. Finally, Omega-3 PUFA preserved sensory and motor function as assessed by two broad-spectrum test batteries. The beneficial effects of Omega-3 PUFA were blocked by an ADAM17 promotor and by a NGF inhibitor, confirming the pathogenic function of ADAM17 and the central neuroprotective role of NGF. Collectively, these findings provide a strong experimental basis for Omega-3 PUFA as a potential clinical treatment for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Ácidos Grasos Omega-3 , Ratones , Animales , Microglía/metabolismo , Factor de Crecimiento Nervioso/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-3/uso terapéutico , Ácidos Grasos Omega-3/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Fenotipo
3.
Nutr Rev ; 81(8): 1051-1062, 2023 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-36409999

RESUMEN

The neurovascular unit (NVU) is composed of neurons, glial cells, and blood vessels. NVU dysfunction involves the processes of neuroinflammation, and microcirculatory disturbances, as well as neuronal injury after traumatic brain injury (TBI). Traditional anti-inflammatory drugs have limited efficacy in improving the prognosis of TBI. Thus, treatments that target NVU dysfunction may provide a breakthrough. A large number of clinical studies have shown that the nutritional status of patients with TBI was closely related to their conditions and prognoses. Nutrient complexes and complementary therapies for the treatment of TBI are therefore being implemented in many preclinical studies. Importantly, the mechanism of action for this treatment may be related to repair of NVU dysfunction by ensuring adequate omega-3 fatty acids, curcumin, resveratrol, apigenin, vitamins, and minerals. These nutritional supplements hold promise for translation to clinical therapy. In addition, dietary habits also play an important role in the rehabilitation of TBI. Poor dietary habits may worsen the pathology and prognosis of TBI. Adjusting dietary habits, especially with a ketogenic diet, may improve outcomes in patients with TBI. This article discusses the impact of clinical nutrition on NVU dysfunction after TBI, focusing on nutritional complexes and dietary habits.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Estado Nutricional , Humanos , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/patología , Suplementos Dietéticos , Microcirculación , Vitaminas/uso terapéutico , Conducta Alimentaria
4.
J Antibiot (Tokyo) ; 72(5): 298-305, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30787402

RESUMEN

Listeria monocytogenes (L. monocytogenes), an important food-borne pathogenic microorganism, has resistance immune function to many commonly used drugs. Myristic acid is a traditional Chinese herbal medicine, but it has been rarely used as a food additive, limiting the development of natural food preservatives. In this study, the antibacterial activity and mechanism of myristic acid against L. monocytogenes were studied. The minimum inhibitory concentration (MIC) of myristic acid against 13 L. monocytogenes strains ranged from 64 to 256 µg ml-1. The time-kill assay demonstrated that when myristic acid was added to dairy products, flow cytometry confirmed that myristic acid influenced cell death and inhibited the growth of L. monocytogenes. Transmission electron microscopy (TEM), scanning electron microscopy (SEM), and NPN uptake studies illustrated that myristic acid changed the bacterial morphology and membrane structure of L. monocytogenes, which led to rapid cell death. Myristic acid could bind to DNA and lead to changes in DNA conformation and structure, as identified by fluorescence spectroscopy. Our studies provide additional evidence to support myristic acid being used as a natural antibacterial agent and also further fundamental understanding of the modes of antibacterial action.


Asunto(s)
Antibacterianos/farmacología , Listeria monocytogenes/efectos de los fármacos , Leche/microbiología , Ácido Mirístico/farmacología , Animales , Membrana Celular/efectos de los fármacos , Transmisión de Enfermedad Infecciosa , Citometría de Flujo , Listeria monocytogenes/citología , Listeria monocytogenes/crecimiento & desarrollo , Listeria monocytogenes/fisiología , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Conformación de Ácido Nucleico/efectos de los fármacos , Espectrometría de Fluorescencia
5.
J Neuroinflammation ; 15(1): 310, 2018 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-30409173

RESUMEN

BACKGROUND: Enhancing autophagy after traumatic brain injury (TBI) may decrease the expression of neuronal apoptosis-related molecules. Autophagy-mediated neuronal survival is regulated by the sirtuin family of proteins (SIRT). Omega-3 polyunsaturated fatty acids (ω-3 PUFA) are known to have antioxidative and anti-inflammatory effects. We previously demonstrated that ω-3 PUFA supplementation attenuated neuronal apoptosis by modulating the neuroinflammatory response through SIRT1-mediated deacetylation of the HMGB1/NF-κB pathway, leading to neuroprotective effects following experimental traumatic brain injury (TBI). However, no studies have elucidated if the neuroprotective effects of ω-3 PUFAs against TBI-induced neuronal apoptosis are modulated by SIRT1-mediated deacetylation of the autophagy pathway. METHODS: The Feeney DM TBI model was adopted to induce TBI rats. Modified neurological severity scores, the rotarod test, brain water content, and Nissl staining were employed to determine the neuroprotective effects of ω-3 PUFA supplementation. Immunofluorescent staining and western blot analysis were used to detect Beclin-1 nuclear translocation and autophagy pathway activation. The impact of SIRT1 deacetylase activity on Beclin-1 acetylation and the interaction between cytoplasmic Beclin-1 and Bcl-2 were assessed to evaluate the neuroprotective effects of ω-3 PUFAs and to determine if these effects were dependent on SIRT1-mediated deacetylation of the autophagy pathway in order to gain further insight into the mechanisms underlying the development of neuroprotection after TBI. RESULTS: ω-3 PUFA supplementation protected neurons against TBI-induced neuronal apoptosis via enhancement of the autophagy pathway. We also found that treatment with ω-3 PUFA significantly increased the NAD+/NADH ratio and SIRT1 activity following TBI. In addition, ω-3 PUFA supplementation increased Beclin-1 deacetylation and its nuclear export and induced direct interactions between cytoplasmic Beclin-1 and Bcl-2 by increasing SIRT1 activity following TBI. These events led to the inhibition of neuronal apoptosis and to neuroprotective effects through enhancing autophagy after TBI, possibly due to elevated SIRT1. CONCLUSIONS: ω-3 PUFA supplementation attenuated TBI-induced neuronal apoptosis by inducing the autophagy pathway through the upregulation of SIRT1-mediated deacetylation of Beclin-1.


Asunto(s)
Apoptosis/efectos de los fármacos , Beclina-1/metabolismo , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-3/uso terapéutico , Sirtuina 1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Autofagia/efectos de los fármacos , Edema Encefálico/etiología , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Hipocampo/citología , Masculino , Enfermedades del Sistema Nervioso/etiología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Prueba de Desempeño de Rotación con Aceleración Constante
6.
Microb Pathog ; 123: 304-313, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30041003

RESUMEN

Staphylococcus aureus (S. aureus) is a Gram-positive bacterium that causes a wide range of diseases, including food poisoning. Tea tree oil (TTO), an essential oil distilled from Melaleuca alternifolia, is well-known for its antibacterial activities. TTO effectively inhibited all 19 tested strains of S. aureus biofilm and planktonic cells. Phenotype analyses of S. aureus biofilm cells exposed to TTO were performed by biofilm adhesion assays, eDNA detection and PIA release. RNA sequencing (RNA-seq) was used in our study to elucidate the mechanism of TTO as a potential antibacterial agent to evaluate differentially expressed genes (DEGs) and the functional network in S. aureus ATCC 29213 biofilms. TTO significantly changed (greater than a 2- or less than a 2-fold change) the expression of 304 genes in S. aureus contained in biofilms. The levels of genes related to the glycine, serine and threonine metabolism pathway, purine metabolism pathway, pyrimidine metabolism pathway and amino acid biosynthesis pathway were dramatically changed in the biofilm exposed to TTO. Furthermore, the expression changes identified by RNA-seq analysis were verified by real-time RT-PCR. To the best of our knowledge, this research is the first study to report the phenotype and expression profiles of S. aureus in biofilms exposed to TTO.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , ARN Bacteriano/análisis , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Aceite de Árbol de Té/farmacología , Aminoácidos/genética , Aminoácidos/metabolismo , Adhesión Bacteriana/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Genes Bacterianos/genética , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/genética , Pruebas de Sensibilidad Microbiana , Fenotipo , Análisis de Secuencia de ARN
7.
J Neuroinflammation ; 15(1): 116, 2018 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-29678169

RESUMEN

BACKGROUND: Microglial polarization and the subsequent neuroinflammatory response are contributing factors for traumatic brain injury (TBI)-induced secondary injury. High mobile group box 1 (HMGB1) mediates the activation of the NF-κB pathway, and it is considered to be pivotal in the late neuroinflammatory response. Activation of the HMGB1/NF-κB pathway is closely related to HMGB1 acetylation, which is regulated by the sirtuin (SIRT) family of proteins. Omega-3 polyunsaturated fatty acids (ω-3 PUFA) are known to have antioxidative and anti-inflammatory effects. We previously demonstrated that ω-3 PUFA inhibited TBI-induced microglial activation and the subsequent neuroinflammatory response by regulating the HMGB1/NF-κB signaling pathway. However, no studies have elucidated if ω-3 PUFA affects the HMGB1/NF-κB pathway in a HMGB1 deacetylation of dependent SIRT1 manner, thus regulating microglial polarization and the subsequent neuroinflammatory response. METHODS: The Feeney DM TBI model was adopted to induce brain injury in rats. Modified neurological severity scores, rotarod test, brain water content, and Nissl staining were employed to determine the neuroprotective effects of ω-3 PUFA supplementation. Assessment of microglia polarization and pro-inflammatory markers, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, and HMGB1, were used to evaluate the neuroinflammatory responses and the anti-inflammatory effects of ω-3 PUFA supplementation. Immunofluorescent staining and western blot analysis were used to detect HMGB1 nuclear translocation, secretion, and HMGB1/NF-κB signaling pathway activation to evaluate the effects of ω-3 PUFA supplementation. The impact of SIRT1 deacetylase activity on HMGB1 acetylation and the interaction between HMGB1 and SIRT1 were assessed to evaluate anti-inflammation effects of ω-3 PUFAs, and also, whether these effects were dependent on a SIRT1-HMGB1/NF-κB axis to gain further insight into the mechanisms underlying the development of the neuroinflammatory response after TBI. RESULTS: The results of our study showed that ω-3 PUFA supplementation promoted a shift from the M1 microglial phenotype to the M2 microglial phenotype and inhibited microglial activation, thus reducing TBI-induced inflammatory factors. In addition, ω-3 PUFA-mediated downregulation of HMGB1 acetylation and its extracellular secretion was found to be likely due to increased SIRT1 activity. We also found that treatment with ω-3 PUFA inhibited HMGB1 acetylation and induced direct interactions between SIRT1 and HMGB1 by elevating SIRT1 activity following TBI. These events lead to inhibition of HMGB1 nucleocytoplasmic translocation/extracellular secretion and alleviated HMGB1-mediated activation of the NF-κB pathway following TBI-induced microglial activation, thus inhibiting the subsequent inflammatory response. CONCLUSIONS: The results of this study suggest that ω-3 PUFA supplementation attenuates the inflammatory response by modulating microglial polarization through SIRT1-mediated deacetylation of the HMGB1/NF-κB pathway, leading to neuroprotective effects following experimental traumatic brain injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/complicaciones , Polaridad Celular/fisiología , Ácidos Grasos Omega-3 , Inflamación/tratamiento farmacológico , Inflamación/etiología , Transducción de Señal/fisiología , Sirtuina 1/metabolismo , Animales , Barrera Hematoencefálica/fisiopatología , Lesiones Traumáticas del Encéfalo/patología , Permeabilidad Capilar/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-3/uso terapéutico , Proteína HMGB1/metabolismo , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Actividad Motora/efectos de los fármacos , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Examen Neurológico , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
8.
Artículo en Chino | WPRIM | ID: wpr-821279

RESUMEN

@#[Abstract] Objective: Toevaluatetheexpressionof6-phosphofructo-2-kinase/fructose-2, 6-biphosphatase 3(PFKFB3) in malignant glioma tissues and the effects of inhibitor of PFKFB3(PFK15) on the proliferation, migration, invasion, clone formation and tumorigenesis of H4 cells. Methods: Malignant brain glioma tissues and corresponding paratumor tissues from 31 patients, who were hospitalized in Department of Neurosurgery,Ankang Hospital of Traditional Chinese Medicine during February 1, 2015 to January 31, 2016 for operative treatment, were collected for this study. Immunohistochemistry and western blotting assays were applied to detect the expression of PFKFB3 in collected tissues. PFKFB3 in H4 cells were blocked by PFK15 (1.25, 2.5, 5.0 μmol/L). The effect of PFK15 on proliferation, migration, clone formation and tumorigenesis of H4 cells were determined by MTT assay, EdU incorporation assay, wound healing assay, Transwell assay, colone formation assay and in vivo xenograft bearing nude mice model respectively. Results: Positive expression rate of PFKFB3 was significantly higher in malignant glioma tissues compared with normal adjacent tissues[(80.60±8.98)% vs (41.57±10.16)%, P<0.05]. The results of MTT assay and EdU incorporation assay indicated that PEK15 significantly inhibited the proliferation of H4 cells in a concentration dependent manner. The migration, invasion and clone formation activity of H4 cells were significantly reduced by treatment with PFK15 (all P<0.05). In tumor bearing nude mice, the tumor volume of mice treated with PFK15 was significantly smaller than that of mice from control group ([254.15±154.25] vs [801.52±224.25] mm3, P<0.05). Conclusion: PFKFB3 was highly expressed in malignant glioma tissues. Blocking of PFKFB3 by PFK15 significantly reduced the malignant biological behaviors and tumorigenesis of H4 cells in vitro and in vivo, which may serve as a promising target for the treatment of malignant gliomas.

9.
J Neuroinflammation ; 14(1): 143, 2017 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-28738820

RESUMEN

BACKGROUND: Microglial activation and the subsequent inflammatory response in the central nervous system play important roles in secondary damage after traumatic brain injury (TBI). High-mobility group box 1 (HMGB1) protein, an important mediator in late inflammatory responses, interacts with transmembrane receptor for advanced glycation end products (RAGE) and toll-like receptors (TLRs) to activate downstream signaling pathways, such as the nuclear factor (NF)-κB signaling pathway, leading to a cascade amplification of inflammatory responses, which are related to neuronal damage after TBI. Omega-3 polyunsaturated fatty acid (ω-3 PUFA) is a commonly used clinical immunonutrient, which has antioxidative and anti-inflammatory effects. However, the effects of ω-3 PUFA on HMGB1 expression and HMGB1-mediated activation of the TLR4/NF-κB signaling pathway are not clear. METHODS: The Feeney DM TBI model was adopted to induce brain injury in rats. Modified neurological severity scores, brain water content, and Nissl staining were employed to determine the neuroprotective effects of ω-3 PUFA supplementation. Assessment of microglial activation in lesioned sites and protein markers for proinflammatory, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, interferon (IFN)-γ, and HMGB1 were used to evaluate neuroinflammatory responses and anti-inflammation effects of ω-3 PUFA supplementation. Immunofluorescent staining and western blot analysis were used to detect HMGB1 nuclear translocation, secretion, and HMGB1-mediated activation of the TLR4/NF-κB signaling pathway to evaluate the effects of ω-3 PUFA supplementation and gain further insight into the mechanisms underlying the development of the neuroinflammatory response after TBI. RESULTS: It was found that ω-3 PUFA supplementation inhibited TBI-induced microglial activation and expression of inflammatory factors (TNF-α, IL-1ß, IL-6, and IFN-γ), reduced brain edema, decreased neuronal apoptosis, and improved neurological functions after TBI. We further demonstrated that ω-3 PUFA supplementation inhibited HMGB1 nuclear translocation and secretion and decreased expression of HMGB1 in neurons and microglia in the lesioned areas. Moreover, ω-3 PUFA supplementation inhibited microglial activation and the subsequent inflammatory response by regulating HMGB1 and the TLR4/NF-κB signaling pathway. CONCLUSIONS: The results of this study suggest that microglial activation and the subsequent neuroinflammatory response as well as the related HMGB1/TLR4/NF-κB signaling pathway play essential roles in secondary injury after TBI. Furthermore, ω-3 PUFA supplementation inhibited TBI-induced microglial activation and the subsequent inflammatory response by regulating HMGB1 nuclear translocation and secretion and also HMGB1-mediated activation of the TLR4/NF-κB signaling pathway, leading to neuroprotective effects.


Asunto(s)
Lesiones Traumáticas del Encéfalo/patología , Encefalitis/dietoterapia , Ácidos Grasos Omega-3/administración & dosificación , Proteína HMGB1/metabolismo , Microglía/efectos de los fármacos , Neuroprostanos/administración & dosificación , Animales , Edema Encefálico/etiología , Lesiones Traumáticas del Encéfalo/complicaciones , Proteínas de Unión al Calcio/metabolismo , Corteza Cerebral/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis/etiología , Encefalitis/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Proteínas de Microfilamentos/metabolismo , Microglía/patología , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Sirtuina 1/metabolismo , Factores de Tiempo
10.
J Ethnopharmacol ; 198: 331-337, 2017 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-28111219

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Licochalcone A (LCA) is a characteristic chalcone that is found in licorice, which is a traditional medicinal plant. In traditional medicine, LCA possesses many potential biological activities, including anti-parasitic, anti-inflammatory and antitumor activities. AIM OF THE STUDY: To determine the antioxidant activity of LCA and, on this basis, to investigate the role of its anticancer activity. MATERIALS AND METHODS: To validate the antioxidant activity of LCA, the proteins SOD, CAT and GPx1 were analyzed using western blotting and cellular antioxidant activity (CAA) assays. Oxidative free radicals are associated with cancer cells. Therefore, the anticancer activity of LCA was also evaluated. To assess the anticancer activity, cell viability assays were performed and apoptosis was evaluated. In addition, MAPK-related proteins were analyzed using western blotting. RESULTS: The experimental data showed that the EC50 of LCA is 58.79±0.05µg/mL and 46.29±0.05µg/mL under the two conditions tested, with or without PBS. In addition, LCA at a concentration of approximately 2-8µg/mL can induce the expression of SOD, CAT and GPx1 proteins. Further, LCA inhibits the growth of HepG2 cells through cell proliferation arrest and the subsequent induction of apoptosis, and LCA attenuated the p38/JNK/ERK signaling pathway in a dose-dependent manner. CONCLUSION: The results showed that LCA suppresses the oxidation of cells and markedly inhibits the proliferation of cancer cells. These findings confirm the traditional use of LCA in folk medicine.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Antioxidantes/farmacología , Chalconas/farmacología , Glycyrrhiza/química , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/aislamiento & purificación , Antioxidantes/administración & dosificación , Antioxidantes/aislamiento & purificación , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Catalasa/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Chalconas/administración & dosificación , Chalconas/aislamiento & purificación , Relación Dosis-Respuesta a Droga , Glutatión Peroxidasa/metabolismo , Células Hep G2 , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Medicina Tradicional/métodos , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa/metabolismo
11.
Bioresour Technol ; 102(3): 2881-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21109424

RESUMEN

The objective of this research was to investigate cellulase adsorption and recycling during enzymatic hydrolysis of two differently pretreated wheat straws (WS). Dilute acid treated WS showed lower hydrolysis yield of polysaccharides fraction and adsorbed more cellulase with hydrolyzed residue than dilute alkali treated sample. Four methods capable of recovering and recycling the enzyme bound to the residual substrate and the enzyme free in solution were used for three consecutive rounds of hydrolysis to compare their recycling efficiencies. Compared to the absorption recycling method, ultrafiltration recycling method possessed the capacity to retain ß-glucosidase, thereby avoiding the supplementation of fresh ß-glucosidase in subsequent rounds of hydrolysis. It was found that whatever recycling method was used, better recycling results were obtained for dilute alkali treated substrate than for dilute acid treated substrate. These results suggested that the great difference in the lignin content between acid treated WS and alkali treated WS would significantly affect enzymatic hydrolysis, cellulase adsorption and cellulase recycling efficiencies.


Asunto(s)
Celulasa/química , Lignina/química , Componentes Aéreos de las Plantas/química , Extractos Vegetales/química , Triticum/química , Ultrafiltración/métodos , Adsorción , Activación Enzimática , Hidrólisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA